Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.392
Filter
1.
J Neurosci Res ; 102(6): e25360, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38847288

ABSTRACT

Childhood obesity increases the risk of health and cognitive disorders in adulthood. Consuming high-fat diets (HFD) during critical neurodevelopmental periods, like childhood, impairs cognition and memory in humans and animals, affecting the function and connectivity of brain structures related to emotional memory. However, the underlying mechanisms of such phenomena need to be better understood. This study aimed to investigate the neurochemical profile of the amygdala and hippocampus, brain structures involved in emotional memory, during the acquisition of conditioned odor aversion in male rats that consumed a HFD from weaning to adulthood. The rats gained weight, experienced metabolic changes, and reduced insulin sensitivity and glucose tolerance. Rats showed enhanced odor aversion memory, contrary to the expected cognitive impairments. This memory enhancement was accompanied by increased noradrenergic and glutamatergic neurotransmission in the amygdala and hippocampus. Importantly, this upregulation was specific to stimuli exposure, as basal neurotransmitter levels remained unaltered by the HFD. Our results suggest that HFD modifies cognitive function by altering neurochemical signaling, in this case, upregulating neurotransmitter levels rendering a stronger memory trace, demonstrating that metabolic dysfunctions do not only trigger exclusively detrimental plasticity processes but also render enhanced plastic effects depending on the type of information.


Subject(s)
Amygdala , Diet, High-Fat , Glutamic Acid , Hippocampus , Synaptic Transmission , Animals , Male , Diet, High-Fat/adverse effects , Hippocampus/metabolism , Amygdala/metabolism , Synaptic Transmission/physiology , Rats , Glutamic Acid/metabolism , Norepinephrine/metabolism , Rats, Wistar , Cognition/physiology , Avoidance Learning/physiology
2.
Sci Rep ; 14(1): 12985, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38839828

ABSTRACT

One third of people with psychosis become antipsychotic treatment-resistant and the underlying mechanisms remain unclear. We investigated whether altered cognitive control function is a factor underlying development of treatment resistance. We studied 50 people with early psychosis at a baseline visit (mean < 2 years illness duration) and follow-up visit (1 year later), when 35 were categorized at treatment-responsive and 15 as treatment-resistant. Participants completed an emotion-yoked reward learning task that requires cognitive control whilst undergoing fMRI and MR spectroscopy to measure glutamate levels from Anterior Cingulate Cortex (ACC). Changes in cognitive control related activity (in prefrontal cortex and ACC) over time were compared between treatment-resistant and treatment-responsive groups and related to glutamate. Compared to treatment-responsive, treatment-resistant participants showed blunted activity in right amygdala (decision phase) and left pallidum (feedback phase) at baseline which increased over time and was accompanied by a decrease in medial Prefrontal Cortex (mPFC) activity (feedback phase) over time. Treatment-responsive participants showed a negative relationship between mPFC activity and glutamate levels at follow-up, no such relationship existed in treatment-resistant participants. Reduced activity in right amygdala and left pallidum at baseline was predictive of treatment resistance at follow-up (67% sensitivity, 94% specificity). The findings suggest that deterioration in mPFC function over time, a key cognitive control region needed to compensate for an initial dysfunction within a social-emotional network, is a factor underlying development of treatment resistance in early psychosis. An uncoupling between glutamate and cognitive control related mPFC function requires further investigation that may present a future target for interventions.


Subject(s)
Cognition , Magnetic Resonance Imaging , Prefrontal Cortex , Psychotic Disorders , Humans , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Prefrontal Cortex/diagnostic imaging , Male , Female , Psychotic Disorders/metabolism , Psychotic Disorders/drug therapy , Psychotic Disorders/physiopathology , Adult , Young Adult , Glutamic Acid/metabolism , Antipsychotic Agents/therapeutic use , Antipsychotic Agents/pharmacology , Gyrus Cinguli/metabolism , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/physiopathology
3.
Nat Commun ; 15(1): 4709, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830891

ABSTRACT

Microbial communities often exhibit more than one possible stable composition for the same set of external conditions. In the human microbiome, these persistent changes in species composition and abundance are associated with health and disease states, but the drivers of these alternative stable states remain unclear. Here we experimentally demonstrate that a cross-kingdom community, composed of six species relevant to the respiratory tract, displays four alternative stable states each dominated by a different species. In pairwise coculture, we observe widespread bistability among species pairs, providing a natural origin for the multistability of the full community. In contrast with the common association between bistability and antagonism, experiments reveal many positive interactions within and between community members. We find that multiple species display cooperative growth, and modeling predicts that this could drive the observed multistability within the community as well as non-canonical pairwise outcomes. A biochemical screening reveals that glutamate either reduces or eliminates cooperativity in the growth of several species, and we confirm that such supplementation reduces the extent of bistability across pairs and reduces multistability in the full community. Our findings provide a mechanistic explanation of how cooperative growth rather than competitive interactions can underlie multistability in microbial communities.


Subject(s)
Microbial Interactions , Microbiota , Microbiota/physiology , Humans , Bacteria/genetics , Bacteria/classification , Bacteria/metabolism , Bacteria/growth & development , Glutamic Acid/metabolism , Models, Biological , Coculture Techniques
4.
Elife ; 122024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829200

ABSTRACT

Threat-response neural circuits are conserved across species and play roles in normal behavior and psychiatric diseases. Maladaptive changes in these neural circuits contribute to stress, mood, and anxiety disorders. Active coping in response to stressors is a psychosocial factor associated with resilience against stress-induced mood and anxiety disorders. The neural circuitry underlying active coping is poorly understood, but the functioning of these circuits could be key for overcoming anxiety and related disorders. The supramammillary nucleus (SuM) has been suggested to be engaged by threat. SuM has many projections and a poorly understood diversity of neural populations. In studies using mice, we identified a unique population of glutamatergic SuM neurons (SuMVGLUT2+::POA) based on projection to the preoptic area of the hypothalamus (POA) and found SuMVGLUT2+::POA neurons have extensive arborizations. SuMVGLUT2+::POA neurons project to brain areas that mediate features of the stress and threat responses including the paraventricular nucleus thalamus (PVT), periaqueductal gray (PAG), and habenula (Hb). Thus, SuMVGLUT2+::POA neurons are positioned as a hub, connecting to areas implicated in regulating stress responses. Here we report SuMVGLUT2+::POA neurons are recruited by diverse threatening stressors, and recruitment correlated with active coping behaviors. We found that selective photoactivation of the SuMVGLUT2+::POA population drove aversion but not anxiety like behaviors. Activation of SuMVGLUT2+::POA neurons in the absence of acute stressors evoked active coping like behaviors and drove instrumental behavior. Also, activation of SuMVGLUT2+::POA neurons was sufficient to convert passive coping strategies to active behaviors during acute stress. In contrast, we found activation of GABAergic (VGAT+) SuM neurons (SuMVGAT+) neurons did not alter drive aversion or active coping, but termination of photostimulation was followed by increased mobility in the forced swim test. These findings establish a new node in stress response circuitry that has projections to many brain areas and evokes flexible active coping behaviors.


Subject(s)
Adaptation, Psychological , Neurons , Stress, Psychological , Animals , Neurons/physiology , Neurons/metabolism , Mice , Adaptation, Psychological/physiology , Male , Glutamic Acid/metabolism , Hypothalamus, Posterior/physiology , Neural Pathways/physiology , Mice, Inbred C57BL
5.
Neurology ; 103(1): e209543, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38870443

ABSTRACT

BACKGROUND AND OBJECTIVES: Cortical lesions contribute to disability in multiple sclerosis (MS), but their impact on regional neurotransmitter levels remains to be clarified. We tested the hypothesis that cortical lesions are associated with regional glutamate and gamma-aminobutyric acid (GABA) concentrations within the affected cortical region. METHODS: In this cross-sectional study, we used structural 7T MRI to segment cortical lesions and 7T proton MR-spectroscopy of the bilateral sensorimotor hand areas to quantify regional GABA, glutamate, N-acetylaspartate, and myoinositol concentrations in patients with MS (inclusion criteria: diagnosis of relapsing-remitting [RR] or secondary progressive MS [SPMS]; age 18-80 years) and age and sex-matched healthy controls. Data were collected at a single center between August 2018 and September 2020. Linear mixed-effects models were used to test for associations between metabolite concentrations and cortical lesion volumes within the same MR-spectroscopy voxel. RESULTS: Forty-seven patients with MS (34 RRMS, 13 SPMS; 45.1 ± 12.5 years; 31 women) and 23 healthy controls (44.4 ± 13 years, 15 women) were studied. In patients, higher regional glutamate and lower regional GABA concentrations were associated with larger cortical lesion volume within the MR-spectroscopy voxel [glutamate: 0.61 (95% CI 0.19-1.03) log(mm3), p = 0.005, GABA: -0.71 (-1.24 to -0.18) log(mm3), p = 0.01]. In addition, lower N-acetylaspartate levels [-0.37 (-0.67 to -0.07) log(mm3), p = 0.016] and higher myoinositol levels [0.48 (0.03-0.93) log(mm3), p = 0.037] were associated with a larger regional cortical lesion volume. Furthermore, glutamate concentrations were reduced in patients with SPMS compared with healthy participants [-0.75 (-1.3 to -0.19) mM, p = 0.005] and patients with RRMS [-0.55 (-1.07 to -0.02) mM, p = 0.04]. N-acetylaspartate levels were lower in both patients with RRMS [-0.81 (-1.39 to -0.24) mM, p = 0.003] and SPMS [-1.31 (-2.07 to -0.54) mM, p < 0.001] when compared with healthy controls. Creatine-normalized N-acetylaspartate levels were associated with performance in the 9-hole peg test of the contralateral hand [-0.004 (-0.007 to -0.002) log(s), p = 0.002], and reduced mean creatine-normalized glutamate was associated with increased Expanded Disability Status Scale (R = -0.39, p = 0.02). DISCUSSION: Cortical lesions are associated with local increases in glutamate and a reduction in GABA concentration within the lesional or perilesional tissue. Further studies are needed to investigate the causal relationship between cortical lesions and changes in neurotransmitter concentrations.


Subject(s)
Aspartic Acid , Cerebral Cortex , Glutamic Acid , Inositol , gamma-Aminobutyric Acid , Humans , Middle Aged , Female , Male , Adult , Inositol/metabolism , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Glutamic Acid/metabolism , gamma-Aminobutyric Acid/metabolism , Cross-Sectional Studies , Cerebral Cortex/metabolism , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Aged , Multiple Sclerosis/metabolism , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Multiple Sclerosis, Chronic Progressive/metabolism , Multiple Sclerosis, Chronic Progressive/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/metabolism , Multiple Sclerosis, Relapsing-Remitting/pathology , Young Adult , Proton Magnetic Resonance Spectroscopy
6.
Eur J Sport Sci ; 24(6): 721-731, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38874966

ABSTRACT

It has been assumed that exercise intensity variation throughout a cycling time trial (TT) occurs in alignment of various metabolic changes to prevent premature task failure. However, this assumption is based on target metabolite responses, which limits our understanding of the complex interconnection of metabolic responses during exercise. The current study characterized the metabolomic profile, an untargeted metabolic analysis, after specific phases of a cycling 4-km TT. Eleven male cyclists performed three separated TTs in a crossover counterbalanced design, which were interrupted at the end of the fast-start (FS, 600 ± 205 m), even-pace (EP, 3600 ± 190 m), or end-spurt (ES, 4000 m) phases. Blood samples were taken before any exercise and 5 min after exercise cessation, and the metabolomic profile characterization was performed using Nuclear Magnetic Resonance metabolomics. Power output (PO) was also continually recorded. There were higher PO values during the FS and ES compared to the EP (all p < 0.05), which were accompanied by distinct metabolomic profiles. FS showed high metabolite expression in TCA cycle and its related pathways (e.g., glutamate, citric acid, and valine metabolism); whereas, the EP elicited changes associated with antioxidant effects and oxygen delivery adjustment. Finally, ES was related to pathways involved in NAD turnover and serotonin metabolism. These findings suggest that the specific phases of a cycling TT are accompanied by distinct metabolomic profiles, providing novel insights regarding the relevance of specific metabolic pathways on the process of exercise intensity regulation.


Subject(s)
Bicycling , Cross-Over Studies , Metabolome , Humans , Male , Metabolome/physiology , Adult , Bicycling/physiology , Citric Acid Cycle , Serotonin/blood , NAD/blood , NAD/metabolism , Young Adult , Glutamic Acid/blood , Glutamic Acid/metabolism , Metabolomics , Valine/blood , Citric Acid/blood
7.
Methods Mol Biol ; 2792: 41-49, 2024.
Article in English | MEDLINE | ID: mdl-38861077

ABSTRACT

Glutamate:glyoxylate aminotransferase (GGAT; EC 2.6.1.4) and serine:glyoxylate aminotransferase activities (SGAT; EC 2.6.1.45) are central photorespiratory reactions within plant peroxisomes. Both enzymatic reactions convert glyoxylate, a product of glycolate oxidase, to glycine, a substrate of the mitochondrial glycine decarboxylase complex. The GGAT reaction uses glutamate as an amino group donor and also produces α-ketoglutarate, which is recycled to glutamate in plastids by ferredoxin-dependent glutamate synthase. Using serine, a product of mitochondrial serine hydroxymethyltransferase, as an amino group donor, the SGAT reaction also produces hydroxypyruvate, a substrate of hydroxypyruvate reductase. The activities of these photorespiratory aminotransferases can be measured using indirect, coupled, spectrophotometric assays, detailed herein.


Subject(s)
Spectrophotometry , Transaminases , Transaminases/metabolism , Spectrophotometry/methods , Glyoxylates/metabolism , Glutamic Acid/metabolism , Enzyme Assays/methods , Cell Respiration
8.
Nat Commun ; 15(1): 4947, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858350

ABSTRACT

The potential brain mechanism underlying resilience to socially transferred allodynia remains unknown. Here, we utilize a well-established socially transferred allodynia paradigm to segregate male mice into pain-susceptible and pain-resilient subgroups. Brain screening results show that ventral tegmental area glutamatergic neurons are selectively activated in pain-resilient mice as compared to control and pain-susceptible mice. Chemogenetic manipulations demonstrate that activation and inhibition of ventral tegmental area glutamatergic neurons bi-directionally regulate resilience to socially transferred allodynia. Moreover, ventral tegmental area glutamatergic neurons that project specifically to the nucleus accumbens shell and lateral habenula regulate the development and maintenance of the pain-resilient phenotype, respectively. Together, we establish an approach to explore individual variations in pain response and identify ventral tegmental area glutamatergic neurons and related downstream circuits as critical targets for resilience to socially transferred allodynia and the development of conceptually innovative analgesics.


Subject(s)
Glutamic Acid , Hyperalgesia , Neurons , Nucleus Accumbens , Ventral Tegmental Area , Animals , Male , Hyperalgesia/physiopathology , Ventral Tegmental Area/physiopathology , Mice , Glutamic Acid/metabolism , Nucleus Accumbens/physiopathology , Neurons/metabolism , Mesencephalon , Mice, Inbred C57BL , Resilience, Psychological , Habenula , Disease Models, Animal
9.
Nat Commun ; 15(1): 4945, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858386

ABSTRACT

Single administration of low-dose ketamine has both acute and sustained anti-depressant effects. Sustained effect is associated with restoration of glutamatergic synapses in medial prefrontal cortic (mFPC) neurons. Ketamine induced profound changes in a number of molecular pathways in a mouse model for chronic stress. Cell-cell communication analyses predicted that planar-cell-polarity (PCP) signaling was decreased after chronic administration of corticosterone but increased following ketamine administration in most of the excitatory neurons. Similar decrease of PCP signaling in excitatory neurons was predicted in dorsolateral prefrontal cortical (dl-PFC) neurons of patients with major depressive disorder (MDD). We showed that the basolateral amygdala (BLA)-projecting infralimbic prefrontal cortex (IL PFC) neurons regulate immobility time in the tail suspension test and food consumption. Conditionally knocking out Celsr2 and Celsr3 or Prickle2 in the BLA-projecting IL PFC neurons abolished ketamine-induced synapse restoration and behavioral remission. Therefore, PCP proteins in IL PFC-BLA neurons mediate synapse restoration induced by of low-dose ketamine.


Subject(s)
Disease Models, Animal , Ketamine , Neurons , Prefrontal Cortex , Synapses , Animals , Ketamine/pharmacology , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Synapses/drug effects , Synapses/metabolism , Neurons/metabolism , Neurons/drug effects , Mice , Male , Humans , Cell Polarity/drug effects , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/drug therapy , Mice, Knockout , Stress, Psychological , Corticosterone , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/drug effects , Mice, Inbred C57BL , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Glutamic Acid/metabolism , Antidepressive Agents/pharmacology
10.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230240, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38853555

ABSTRACT

Synaptic plasticity is a key cellular model for learning, memory and chronic pain. Most previous studies were carried out in rats and mice, and less is known about synaptic plasticity in non-human primates. In the present study, we used integrative experimental approaches to study long-term potentiation (LTP) in the anterior cingulate cortex (ACC) of adult tree shrews. We found that glutamate is the major excitatory transmitter and α-amino-3-hydroxy-5-methyl-4-isoxazole-propionicacid (AMPA) receptors mediate postsynaptic responses. LTP in tree shrews was greater than that in adult mice and lasted for at least 5 h. N-methyl-d-aspartic acid (NMDA) receptors, Ca2+ influx and adenylyl cyclase 1 (AC1) contributed to tree shrew LTP. Our results suggest that LTP is a major form of synaptic plasticity in the ACC of primate-like animals. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Subject(s)
Gyrus Cinguli , Long-Term Potentiation , Receptors, AMPA , Receptors, N-Methyl-D-Aspartate , Tupaiidae , Animals , Long-Term Potentiation/physiology , Gyrus Cinguli/physiology , Tupaiidae/physiology , Mice , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, AMPA/metabolism , Adenylyl Cyclases/metabolism , Glutamic Acid/metabolism , Male
11.
Acta Neuropathol ; 147(1): 98, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861157

ABSTRACT

Widespread cortical accumulation of misfolded pathological tau proteins (ptau) in the form of paired helical filaments is a major hallmark of Alzheimer's disease. Subcellular localization of ptau at various stages of disease progression is likely to be informative of the cellular mechanisms involving its spread. Here, we found that the density of ptau within several distinct rostral thalamic nuclei in post-mortem human tissue (n = 25 cases) increased with the disease stage, with the anterodorsal nucleus (ADn) consistently being the most affected. In the ADn, ptau-positive elements were present already in the pre-cortical (Braak 0) stage. Tau pathology preferentially affected the calretinin-expressing subpopulation of glutamatergic neurons in the ADn. At the subcellular level, we detected ptau immunoreactivity in ADn cell bodies, dendrites, and in a specialized type of presynaptic terminal that expresses vesicular glutamate transporter 2 (vGLUT2) and likely originates from the mammillary body. The ptau-containing terminals displayed signs of degeneration, including endosomal/lysosomal organelles. In contrast, corticothalamic axon terminals lacked ptau. The data demonstrate the involvement of a specific cell population in ADn at the onset of the disease. The presence of ptau in subcortical glutamatergic presynaptic terminals supports hypotheses about the transsynaptic spread of tau selectively affecting specialized axonal pathways.


Subject(s)
Alzheimer Disease , tau Proteins , Humans , tau Proteins/metabolism , Female , Male , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Middle Aged , Neurons/metabolism , Neurons/pathology , Vesicular Glutamate Transport Protein 2/metabolism , Glutamic Acid/metabolism , Anterior Thalamic Nuclei/metabolism , Anterior Thalamic Nuclei/pathology , Calbindin 2/metabolism , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology
12.
Nat Commun ; 15(1): 4537, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806470

ABSTRACT

The multidrug efflux transporter EmrE from Escherichia coli requires anionic residues in the substrate binding pocket for coupling drug transport with the proton motive force. Here, we show how protonation of a single membrane embedded glutamate residue (Glu14) within the homodimer of EmrE modulates the structure and dynamics in an allosteric manner using NMR spectroscopy. The structure of EmrE in the Glu14 protonated state displays a partially occluded conformation that is inaccessible for drug binding by the presence of aromatic residues in the binding pocket. Deprotonation of a single Glu14 residue in one monomer induces an equilibrium shift toward the open state by altering its side chain position and that of a nearby tryptophan residue. This structural change promotes an open conformation that facilitates drug binding through a conformational selection mechanism and increases the binding affinity by approximately 2000-fold. The prevalence of proton-coupled exchange in efflux systems suggests a mechanism that may be shared in other antiporters where acid/base chemistry modulates access of drugs to the substrate binding pocket.


Subject(s)
Antiporters , Escherichia coli Proteins , Escherichia coli , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Antiporters/metabolism , Antiporters/chemistry , Antiporters/genetics , Binding Sites , Protein Binding , Protons , Protein Conformation , Magnetic Resonance Spectroscopy , Glutamic Acid/metabolism , Glutamic Acid/chemistry , Models, Molecular
13.
Cell Rep ; 43(5): 114186, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38700985

ABSTRACT

The fine control of synaptic function requires robust trans-synaptic molecular interactions. However, it remains poorly understood how trans-synaptic bridges change to reflect the functional states of the synapse. Here, we develop optical tools to visualize in firing synapses the molecular behavior of two trans-synaptic proteins, LGI1 and ADAM23, and find that neuronal activity acutely rearranges their abundance at the synaptic cleft. Surprisingly, synaptic LGI1 is primarily not secreted, as described elsewhere, but exo- and endocytosed through its interaction with ADAM23. Activity-driven translocation of LGI1 facilitates the formation of trans-synaptic connections proportionally to the history of activity of the synapse, adjusting excitatory transmission to synaptic firing rates. Accordingly, we find that patient-derived autoantibodies against LGI1 reduce its surface fraction and cause increased glutamate release. Our findings suggest that LGI1 abundance at the synaptic cleft can be acutely remodeled and serves as a critical control point for synaptic function.


Subject(s)
Intracellular Signaling Peptides and Proteins , Synapses , Synaptic Transmission , Synaptic Transmission/physiology , Humans , Synapses/metabolism , Animals , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Glutamic Acid/metabolism , Protein Transport , Male , ADAM Proteins/metabolism , Neurons/metabolism , Autoantibodies/immunology , Mice, Inbred C57BL
14.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38755010

ABSTRACT

Cholinergic neurons of the basal forebrain represent the main source of cholinergic innervation of large parts of the neocortex and are involved in adults in the modulation of attention, memory, and arousal. During the first postnatal days, they play a crucial role in the development of cortical neurons and cortical cytoarchitecture. However, their characteristics, during this period have not been studied. To understand how they can fulfill this role, we investigated the morphological and electrophysiological maturation of cholinergic neurons of the substantia innominata-nucleus basalis of Meynert (SI/NBM) complex in the perinatal period in mice. We show that cholinergic neurons, whether or not they express gamma-aminobutyric acid (GABA) as a cotransmitter, are already functional at Embryonic Day 18. Until the end of the first postnatal week, they constitute a single population of neurons with a well developed dendritic tree, a spontaneous activity including bursting periods, and a short-latency response to depolarizations (early-firing). They are excited by both their GABAergic and glutamatergic afferents. During the second postnatal week, a second, less excitable, neuronal population emerges, with a longer delay response to depolarizations (late-firing), together with the hyperpolarizing action of GABAA receptor-mediated currents. This classification into early-firing (40%) and late-firing (60%) neurons is again independent of the coexpression of GABAergic markers. These results strongly suggest that during the first postnatal week, the specific properties of developing SI/NBM cholinergic neurons allow them to spontaneously release acetylcholine (ACh), or ACh and GABA, into the developing cortex.


Subject(s)
Basal Forebrain , Cholinergic Neurons , gamma-Aminobutyric Acid , Animals , Cholinergic Neurons/physiology , Cholinergic Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Basal Forebrain/physiology , Basal Forebrain/metabolism , Animals, Newborn , Mice, Inbred C57BL , Female , Basal Nucleus of Meynert/physiology , Basal Nucleus of Meynert/metabolism , Substantia Innominata/physiology , Substantia Innominata/metabolism , Mice , Receptors, GABA-A/metabolism , Action Potentials/physiology , Patch-Clamp Techniques , Glutamic Acid/metabolism
15.
J Chem Inf Model ; 64(10): 4168-4179, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38745447

ABSTRACT

Hydroxyprolines are abundant in nature and widely utilized by many living organisms. Isomerization of trans-4-hydroxy-d-proline (t4D-HP) to generate 2-amino-4-ketopentanoate has been found to need a glycyl radical enzyme HplG, which catalyzes the cleavage of the C-N bond, while dehydration of trans-4-hydroxy-l-proline involves a homologous enzyme of HplG. Herein, molecular dynamics simulations and quantum mechanics/molecular mechanics (QM/MM) calculations are employed to understand the reaction mechanism of HplG. Two possible reaction pathways of HplG have been explored to decipher the origin of its chemoselectivity. The QM/MM calculations reveal that the isomerization proceeds via an initial hydrogen shift from the Cγ site of t4D-HP to a catalytic cysteine radical, followed by cleavage of the Cδ-N bond in t4D-HP to form a radical intermediate that captures a hydrogen atom from the cysteine. Activation of the Cδ-H bond in t4D-HP to bring about dehydration of t4D-HP possesses an extremely high energy barrier, thus rendering the dehydration pathway implausible in HplG. On the basis of the current calculations, conserved residue Glu429 plays a pivotal role in the isomerization pathway: the hydrogen bonding between it and t4D-HP weakens the hydroxyalkyl Cγ-Hγ bond, and it acts as a proton acceptor to trigger the cleavage of the C-N bond in t4D-HP. Our current QM/MM calculations rationalize the origin of the experimentally observed chemoselectivity of HplG and propose an H-bond-assisted bond activation strategy in radical-containing enzymes. These findings have general implications on radical-mediated enzymatic catalysis and expand our understanding of how nature wisely and selectively activates the C-H bond to modulate catalytic selectivity.


Subject(s)
Cysteine , Glutamic Acid , Molecular Dynamics Simulation , Quantum Theory , Cysteine/chemistry , Cysteine/metabolism , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Free Radicals/chemistry , Free Radicals/metabolism , Hydrogen Bonding
16.
PLoS One ; 19(5): e0303235, 2024.
Article in English | MEDLINE | ID: mdl-38728287

ABSTRACT

Excitotoxicity represents the primary cause of neuronal death following spinal cord injury (SCI). While autophagy plays a critical and intricate role in SCI, the specific mechanism underlying the relationship between excitotoxicity and autophagy in SCI has been largely overlooked. In this study, we isolated primary spinal cord neurons from neonatal rats and induced excitotoxic neuronal injury by high concentrations of glutamic acid, mimicking an excitotoxic injury model. Subsequently, we performed transcriptome sequencing. Leveraging machine learning algorithms, including weighted correlation network analysis (WGCNA), random forest analysis (RF), and least absolute shrinkage and selection operator analysis (LASSO), we conducted a comprehensive investigation into key genes associated with spinal cord neuron injury. We also utilized protein-protein interaction network (PPI) analysis to identify pivotal proteins regulating key gene expression and analyzed key genes from public datasets (GSE2599, GSE20907, GSE45006, and GSE174549). Our findings revealed that six genes-Anxa2, S100a10, Ccng1, Timp1, Hspb1, and Lgals3-were significantly upregulated not only in vitro in neurons subjected to excitotoxic injury but also in rats with subacute SCI. Furthermore, Hspb1 and Lgals3 were closely linked to neuronal autophagy induced by excitotoxicity. Our findings contribute to a better understanding of excitotoxicity and autophagy, offering potential targets and a theoretical foundation for SCI diagnosis and treatment.


Subject(s)
Autophagy , Galectin 3 , Machine Learning , Neurons , Animals , Rats , Galectin 3/metabolism , Galectin 3/genetics , Glutamic Acid/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neurons/metabolism , Protein Interaction Maps , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/genetics
18.
Nat Commun ; 15(1): 4549, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811525

ABSTRACT

Breast cancer metastasis to the brain is a clinical challenge rising in prevalence. However, the underlying mechanisms, especially how cancer cells adapt a distant brain niche to facilitate colonization, remain poorly understood. A unique metabolic feature of the brain is the coupling between neurons and astrocytes through glutamate, glutamine, and lactate. Here we show that extracellular vesicles from breast cancer cells with a high potential to develop brain metastases carry high levels of miR-199b-5p, which shows higher levels in the blood of breast cancer patients with brain metastases comparing to those with metastatic cancer in other organs. miR-199b-5p targets solute carrier transporters (SLC1A2/EAAT2 in astrocytes and SLC38A2/SNAT2 and SLC16A7/MCT2 in neurons) to hijack the neuron-astrocyte metabolic coupling, leading to extracellular retention of these metabolites and promoting cancer cell growth. Our findings reveal a mechanism through which cancer cells of a non-brain origin reprogram neural metabolism to fuel brain metastases.


Subject(s)
Astrocytes , Brain Neoplasms , Breast Neoplasms , MicroRNAs , Neurons , Humans , MicroRNAs/metabolism , MicroRNAs/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Brain Neoplasms/secondary , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Female , Animals , Cell Line, Tumor , Astrocytes/metabolism , Astrocytes/pathology , Neurons/metabolism , Neurons/pathology , Mice , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 2/genetics , Extracellular Vesicles/metabolism , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/genetics , Gene Expression Regulation, Neoplastic , Glutamic Acid/metabolism , Glutamine/metabolism , Brain/metabolism , Brain/pathology , Lactic Acid/metabolism , Cell Proliferation
19.
Proc Natl Acad Sci U S A ; 121(23): e2318843121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38805277

ABSTRACT

The development and performance of two mass spectrometry (MS) workflows for the intraoperative diagnosis of isocitrate dehydrogenase (IDH) mutations in glioma is implemented by independent teams at Mayo Clinic, Jacksonville, and Huashan Hospital, Shanghai. The infiltrative nature of gliomas makes rapid diagnosis necessary to guide the extent of surgical resection of central nervous system (CNS) tumors. The combination of tissue biopsy and MS analysis used here satisfies this requirement. The key feature of both described methods is the use of tandem MS to measure the oncometabolite 2-hydroxyglutarate (2HG) relative to endogenous glutamate (Glu) to characterize the presence of mutant tumor. The experiments i) provide IDH mutation status for individual patients and ii) demonstrate a strong correlation of 2HG signals with tumor infiltration. The measured ratio of 2HG to Glu correlates with IDH-mutant (IDH-mut) glioma (P < 0.0001) in the tumor core data of both teams. Despite using different ionization methods and different mass spectrometers, comparable performance in determining IDH mutations from core tumor biopsies was achieved with sensitivities, specificities, and accuracies all at 100%. None of the 31 patients at Mayo Clinic or the 74 patients at Huashan Hospital were misclassified when analyzing tumor core biopsies. Robustness of the methodology was evaluated by postoperative re-examination of samples. Both teams noted the presence of high concentrations of 2HG at surgical margins, supporting future use of intraoperative MS to monitor for clean surgical margins. The power of MS diagnostics is shown in resolving contradictory clinical features, e.g., in distinguishing gliosis from IDH-mut glioma.


Subject(s)
Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Mutation , Glioma/genetics , Glioma/surgery , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Humans , Brain Neoplasms/genetics , Brain Neoplasms/surgery , Brain Neoplasms/pathology , Tandem Mass Spectrometry/methods , Glutarates/metabolism , Mass Spectrometry/methods , Glutamic Acid/metabolism , Glutamic Acid/genetics
20.
Chronobiol Int ; 41(6): 802-816, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38757583

ABSTRACT

Stable and entrainable physiological circadian rhythms are crucial for overall health and well-being. The suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals, consists of diverse neuron types that collectively generate a circadian profile of electrical activity. However, the mechanisms underlying the regulation of endogenous neuronal excitability in the SCN remain unclear. Two-pore domain potassium channels (K2P), including TASK-3, are known to play a significant role in maintaining SCN diurnal homeostasis by inhibiting neuronal activity at night. In this study, we investigated the role of TASK-3 in SCN circadian neuronal regulation and behavioural photoentrainment using a TASK-3 global knockout mouse model. Our findings demonstrate the importance of TASK-3 in maintaining SCN hyperpolarization during the night and establishing SCN sensitivity to glutamate. Specifically, we observed that TASK-3 knockout mice lacked diurnal variation in resting membrane potential and exhibited altered glutamate sensitivity both in vivo and in vitro. Interestingly, despite these changes, the mice lacking TASK-3 were still able to maintain relatively normal circadian behaviour.


Subject(s)
Circadian Rhythm , Mice, Knockout , Potassium Channels, Tandem Pore Domain , Suprachiasmatic Nucleus , Animals , Circadian Rhythm/physiology , Suprachiasmatic Nucleus/physiology , Suprachiasmatic Nucleus/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Mice , Male , Mice, Inbred C57BL , Behavior, Animal/physiology , Glutamic Acid/metabolism , Neurons/physiology , Neurons/metabolism , Membrane Potentials/physiology , Potassium Channels
SELECTION OF CITATIONS
SEARCH DETAIL
...